1
|
Liu G, Jia R, Deng H, Wang X. Syndecan-4 promotes gastric cancer progression through activating TGF-β1 induced lipid reprogramming and contributes positive loop circuits. Discov Oncol 2025; 16:1104. [PMID: 40516011 DOI: 10.1007/s12672-025-02975-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND Syndecan-4 (SDC4) plays a pivotal role in tumorigenesis through cell signaling, adhesion and matrix interactions. Lipid reprogramming is a core strategy for malignant tumors to drive progression and invasion through reshaping immunity, stromal and vascular ecology in microenvironment. However, the regulatory mechanism of SDC4 in lipid reprogramming has not yet been elucidated. Therefore, it is important to investigate underlying mechanisms and develop novel therapeutic targets for gastric cancer (GC). METHODS Clinicopathological data and corresponding immunohistochemistry were collected to explore the role of SDC4 in patients with GC. Functional experiments were performed to assess tumor progression and lipid reprogramming. Moreover, western blot assay was performed to verify the molecular mechanisms. In addition, cholesterol-induced lipotoxic environments both in vivo and in vitro were constructed to explore the underlying positive loop circuit. RESULTS SDC4 expression was upregulated in tumor tissues compared to normal gastric tissues and was associated with differentiation grades. Patients with high SDC4 expression were positively correlated with high circulating tumor cell (CTC) levels, vascular endothelial growth factor (VEGF) levels and poor prognosis. Moreover, SDC4 significantly promoted tumor progression by activating transforming growth factor-beta 1 (TGF-β1/TGFB1)-induced lipid reprogramming and contributed a positive loop circuit in GC cells. However, the core of this circuit was dependent on Smad3. In addition, a cholesterol-induced lipotoxic environment upregulated SDC4 expression by activating the RAS signaling pathway and further indicated a positive loop circuit between SDC4 and lipid reprogramming in GC. CONCLUSIONS These findings highlight SDC4 as a therapeutic target for GC and identify actionable positive loop circuits.
Collapse
Affiliation(s)
- Guangjie Liu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ru Jia
- Department of Anorectal Surgery, Anhui Acupuncture and Moxibustion Clinical Medicine Research Center, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, Anhui, China
| | - Heng Deng
- Department of Anorectal Surgery, Anhui Acupuncture and Moxibustion Clinical Medicine Research Center, Second Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, 230000, Anhui, China.
| | - Xiaoshan Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
2
|
Solomon MA, Hains DS, Schwaderer AL, Gallaway K, Sallee CJ, Pike F, Arregui S, Cater DT, Mastropietro CW, Rowan CM. Syndecan-1 as a Biomarker for Fluid Overload After High-Risk Pediatric Cardiac Surgery: A Pilot Study. Pediatr Crit Care Med 2025; 26:e622-e632. [PMID: 40062810 PMCID: PMC12061566 DOI: 10.1097/pcc.0000000000003717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Fluid overload (FO) after pediatric cardiac surgery with cardiopulmonary bypass (CPB) is common and has been associated with poor outcomes. We aimed to describe the relationship between plasma concentrations of syndecan-1 (SD1), a biomarker of endothelial glycocalyx injury, and FO in a cohort of children undergoing cardiac surgery. DESIGN Single-center prospective observational pilot study, 2022-2023. SETTING Twenty-six-bed pediatric cardiac ICU (CICU) at a quaternary pediatric referral center. PATIENTS Children younger than 18 years old undergoing Society of Thoracic Surgeons-European Association for Cardio-Thoracic Surgery congenital heart surgery mortality category 3, 4, and 5 cardiac surgeries with CPB. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS We enrolled 15 patients. Blood samples were collected preoperatively and 4 hours postoperatively, then processed for plasma. SD1 concentrations were measured using enzyme-linked immunosorbent assays and compared with fluid balance on postoperative days (PODs) 1, 2, 3, and peak. SD1 discriminated fluid balance of greater than or equal to 10% on POD-1, POD-2, and POD-3 with an area under the receiver operating characteristic curve (AUROC) of 0.74, 0.84, and 0.88, respectively. SD1 also discriminated peak fluid balance of greater than or equal to 10% occurring on any day over the first seven PODs with an AUROC of 0.94. Patients with greater than or equal to 10% fluid balance on POD-2 ( p = 0.037), POD-3 ( p = 0.020), or peak ( p = 0.021) had significantly elevated delta SD1 when compared with those reaching less than 10%. Fluid balance of greater than or equal to 10% on POD-2 was associated with adverse events including longer duration of mechanical ventilation and CICU stay. CONCLUSIONS Plasma SD1 was associated with FO in pediatric patients undergoing high-risk cardiac surgery with CPB. Further studies exploring the clinical utility of SD1 as a biomarker for FO in the postoperative management of children who undergo cardiac surgery with CPB should be pursued.
Collapse
Affiliation(s)
- Matthew A Solomon
- Division of Pediatric Critical Care, Riley Hospital for Children, Indianapolis, IN
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - David S Hains
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Division of Pediatric Nephrology, Riley Hospital for Children, Indianapolis, IN
| | - Andrew L Schwaderer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
- Division of Pediatric Nephrology, Riley Hospital for Children, Indianapolis, IN
| | - Katie Gallaway
- Division of Pediatric Critical Care, Riley Hospital for Children, Indianapolis, IN
| | - Colin J Sallee
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA
| | - Francis Pike
- Department of Biostatistics and Data Science, Indiana University, Indianapolis, IN
| | - Sam Arregui
- Division of Pediatric Nephrology, Riley Hospital for Children, Indianapolis, IN
| | - Daniel T Cater
- Division of Pediatric Critical Care, Riley Hospital for Children, Indianapolis, IN
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Christopher W Mastropietro
- Division of Pediatric Critical Care, Riley Hospital for Children, Indianapolis, IN
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Courtney M Rowan
- Division of Pediatric Critical Care, Riley Hospital for Children, Indianapolis, IN
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
3
|
Pang J, Xu D, Zhang X, Qu J, Jiang J, Suo J, Li T, Li Y, Peng Z. TIMP2-mediated mitochondrial fragmentation and glycolytic reprogramming drive renal fibrogenesis following ischemia-reperfusion injury. Free Radic Biol Med 2025; 232:244-259. [PMID: 39986488 DOI: 10.1016/j.freeradbiomed.2025.02.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Acute kidney injury (AKI) triggers renal structural and functional abnormalities through inflammatory and fibrotic signaling pathways, ultimately progressing to chronic kidney disease (CKD). The mechanisms underlying AKI-to-CKD transition are complex, with hypoxia, mitochondrial dysfunction, and metabolic reprogramming as critical contributors. Public data analysis demonstrated significant upregulation of tissue inhibitors of metalloproteinases (Timp2) in renal biopsy tissues of CKD patients. In both ischemia/reperfusion (I/R) and unilateral ureteral obstruction (UUO) models, Timp2 upregulation was observed. Tubule-specific Timp2 knockout markedly attenuated renal fibrosis. RNA-sequencing revealed Timp2's association with mitochondrial dynamics and glycolysis in I/R mice. Timp2 deletion improved mitochondrial morphology and suppressed glycolytic enzyme expression. In vitro, TGF-β1-treated Timp2-knockdown HK-2 cells exhibited inhibited Drp1 expression, restored Mfn2 levels, alleviated mitochondrial fragmentation, and elevated mitochondrial membrane potential. Additionally, Pfkfb3 and HIF-1α were downregulated, accompanied by reduced extracellular acidification rate (ECAR), PFK activity, and lactate production. Mechanistically, Timp2 interacts with the extracellular domain of Sdc4 in an autocrine manner, activating the Hedgehog (Hh) signaling pathway. Cyclopamine partially rescued Timp2 overexpression-induced mitochondrial dysfunction, suppressed Pfkfb3-mediated glycolysis, and diminished collagen deposition. This study is the first to demonstrate that Timp2 in TECs exacerbates Hh signaling, promoting mitochondrial fragmentation and metabolic reprogramming to accelerate I/R-induced renal fibrosis.
Collapse
Affiliation(s)
- Jingjing Pang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Dongxue Xu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Xiaoyu Zhang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jiacheng Qu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jun Jiang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Jinmeng Suo
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Tianlong Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Yiming Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China; Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China.
| | - Zhiyong Peng
- Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China; Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Intensive Care Unit of the Second Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China.
| |
Collapse
|
4
|
He K, Ren H, Chen X, He F, Zhang Y, Zhang H, Li F, Yu S. Shed Syndecan-4 and Its Possible Roles in Osteoarthritis. Biomedicines 2025; 13:1037. [PMID: 40426865 PMCID: PMC12109450 DOI: 10.3390/biomedicines13051037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
The specific pathogenesis of osteoarthritis (OA) remains not fully understood. As a transmembrane heparan sulfate proteoglycan, syndecan-4 (SDC4) has been proven to play an important role in the development of OA. Notably, the extracellular domain of SDC4 can be cleaved by proteolytic enzymes, leading to the release of shed SDC4 (sSDC4), which subsequently regulates various biological processes in an autocrine or paracrine manner. This review analyzed 97 publications (1987-2025) from Pubmed and the Web of Science Core Collection using specific key words (syndecan-4, shed syndecan-4, and osteoarthritis), providing a comprehensive overview of the current research on sSDC4, including its shedding enzymes and specific cleavage sites, as well as the factors and mechanisms that influence SDC4 shedding. Furthermore, it summarizes the functions of both sSDC4 and its remaining membrane-bound domain. Finally, the roles of sSDC4 in OA are discussed to identify potential therapeutic targets and explore new strategies for the treatment of OA.
Collapse
Affiliation(s)
- Kangping He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Haozhe Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Xiaohua Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Feng He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Yueying Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Department of Orthodontics, College of Stomatology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Hongyun Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Feifei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Shibin Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
5
|
Hudák A, Letoha T. Endocytic Pathways Unveil the Role of Syndecans in the Seeding and Spreading of Pathological Protein Aggregates: Insights into Neurodegenerative Disorders. Int J Mol Sci 2025; 26:4037. [PMID: 40362276 PMCID: PMC12071627 DOI: 10.3390/ijms26094037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease and other neurodegenerative disorders are characterized by the accumulation of misfolded proteins, such as amyloid-beta, tau, and α-synuclein, which disrupt neuronal function and contribute to cognitive decline. Heparan sulfate proteoglycans, particularly syndecans, play a pivotal role in the seeding, aggregation, and spreading of toxic protein aggregates through endocytic pathways. Among these, syndecan-3 is particularly critical in regulating the internalization of misfolded proteins, facilitating their propagation in a prion-like manner. This review examines the mechanisms by which syndecans, especially SDC3, contribute to the seeding and spreading of pathological protein aggregates in neurodegenerative diseases. Understanding these endocytic pathways provides valuable insights into the potential of syndecans as biomarkers and therapeutic targets for early intervention in Alzheimer's disease and other related neurodegenerative disorders.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
6
|
Hakami H, Dinesh NEH, Nelea V, Lamarche‐Vane N, Ricard‐Blum S, Reinhardt DP. Fibulin-4 and latent-transforming growth factor beta-binding protein-4 interactions with syndecan-2 and syndecan-3 are required for elastogenesis. FASEB J 2025; 39:e70505. [PMID: 40168061 PMCID: PMC11960800 DOI: 10.1096/fj.202402767r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Elastogenesis is a cell surface-located hierarchical process that requires the core components tropoelastin and fibrillins and several accessory proteins, including fibulin-4 (FBLN4) and latent TGF-β binding protein-4 (LTBP4). FBLN4 and LTBP4 interact with cells, but their cell receptors and associated molecular elastogenic mechanisms remain unknown. Primary skin fibroblasts and several vascular smooth muscle cells bound strongly to FBLN4 multimers and LTBP4 monomers. We identified two cell interaction epitopes on FBLN4 located in cbEGF2-3 and the C-terminal domain, whereas FBLN4 multimerization sites were mapped to cbEGF4-5 and the C-terminal domain. We also determined a novel cell interaction site in the N-terminal half of LTBP4. Cell binding to FBLN4 and LTBP4 was strongly inhibited in the presence of heparin, heparan sulfate, or after enzymatic removal of heparan sulfate, suggesting heparan sulfate proteoglycans as relevant cell surface receptors. siRNA knockdown experiments identified syndecan (SDC)2 and SDC3 as cell receptors for FBNL4 and SDC3 for LTBP4. Direct protein interactions between FBLN4 and the recombinant ectodomains of SDC2 and SDC3, and between LTBP4 and SDC3 validated these results. Interaction of the elastogenic cells with FBLN4 and LTBP4 enhanced elastogenesis, whereas SDC2 and/or SDC3 knockdowns led to reduced elastic fiber formation. The cell interactions with FBLN4 and LTBP4 significantly enhanced focal adhesion formation, induced cell contraction, and led to activation of focal adhesion kinase (FAK), Erk1/2, and RhoA. Pharmacological inhibition of these effectors markedly attenuated elastic fiber formation, and siRNA knockdown of SDC2 and SDC3 led to reduced levels of pFAK, pERK, and active RhoA. Together, these data demonstrate that FBLN4 and LTBP4 cell interactions through SDC2 and SDC3 promote elastogenesis by enhancing focal adhesion formation, leading to cell contractility through FAK, Erk1/2, and RhoA activation, underscoring the significance of these pathways in elastogenesis.
Collapse
Affiliation(s)
- Hana Hakami
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell BiologyMcGill UniversityMontréalQuebecCanada
- Faculty of Sciences and Medical Studies, College of Sciences, Zoology DepartmentKing Saud UniversityRiyadhSaudi Arabia
| | - Neha E. H. Dinesh
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell BiologyMcGill UniversityMontréalQuebecCanada
| | - Valentin Nelea
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell BiologyMcGill UniversityMontréalQuebecCanada
- Faculty of Dental Medicine and Oral Health SciencesMcGill UniversityMontréalQuebecCanada
| | - Nathalie Lamarche‐Vane
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell BiologyMcGill UniversityMontréalQuebecCanada
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontréalQuebecCanada
| | - Sylvie Ricard‐Blum
- Institute of Molecular and Supramolecular Chemistry and Biochemistry (ICBMS)UMR 5246, CNRS, University Claude Bernard Lyon 1VilleurbanneFrance
| | - Dieter P. Reinhardt
- Faculty of Medicine and Health Sciences, Department of Anatomy and Cell BiologyMcGill UniversityMontréalQuebecCanada
- Cancer Research ProgramResearch Institute of the McGill University Health CentreMontréalQuebecCanada
| |
Collapse
|
7
|
Guyot E. Heparan sulfate chains in hepatocellular carcinoma. Gastroenterol Rep (Oxf) 2025; 13:goaf023. [PMID: 40093586 PMCID: PMC11908768 DOI: 10.1093/gastro/goaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/13/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Hepatocellular carcinoma (HCC) corresponds to the vast majority of liver cancer cases, with one of the highest mortality rates. Major advances have been made in this field both in the characterization of the molecular pathogenesis and in the development of systemic therapies. Despite these achievements, biomarkers and more efficient treatments are still needed to improve its management. Heparan sulfate (HS) chains are polysaccharides that are present at the cell surface or in the extracellular matrix that are able to bind various types of molecules, such as soluble factors, affecting their availability and thus their effects, or to contribute to interactions that position cells in their environments. Enzymes can modify HS chains after their synthesis, thus changing their properties. Numerous studies have shown HS-related proteins to be key actors that are associated with cellular effects, such as tumor growth, invasion, and metastasis, including in the context of liver carcinogenesis. The aim of this review is to provide a comprehensive overview of the biology of HS chains and their potential importance in HCC, from biological considerations to clinical development, and the identification of biomarkers, as well as therapeutic perspectives.
Collapse
Affiliation(s)
- Erwan Guyot
- Biochemistry Unit, Saint-Antoine Hospital, AP-HP Sorbonne University, Paris Cedex, France
| |
Collapse
|
8
|
Lai SK, Lee ZQ, Tan TI, Tan BH, Sugrue RJ. Evidence that the cell glycocalyx envelops respiratory syncytial virus (RSV) particles that form on the surface of RSV-infected human airway cells. Virology 2025; 604:110415. [PMID: 40044247 DOI: 10.1016/j.virol.2025.110415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/29/2024] [Accepted: 01/16/2025] [Indexed: 05/11/2025]
Abstract
We examined how respiratory syncytial virus (RSV) particles circumvent the overlying glycocalyx on virus-infected A549 cells. The glycocalyx was detected using the lectin WGA-AL488 probe, and the antibodies anti-HS and anti-syndecan-4 that detect heparin sulphate (HS) and the syndecan-4 protein (SYND4) respectively. Imaging of RSV-infected cells provided evidence that the glycocalyx envelopes the virus filaments as they form, and that components of the glycocalyx such as HS moieties and SYND4 are displayed on the surface of the mature virus filaments. Recombinant expression of the G protein in these cells suggested that the G protein was trafficked into pre-existing filamentous cellular structures with a well-defined glycocalyx, further suggesting that the glycocalyx is maintained at the site of virus particle assembly. These data provide evidence that during RSV particle assembly the virus filaments become enveloped by the glycocalyx, and that the glycocalyx should be considered as a structural component of virus filaments.
Collapse
Affiliation(s)
- Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Zhi Qi Lee
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Trina Isabel Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Boon Huan Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.
| |
Collapse
|
9
|
Arokiasamy S, Balderstone MJM, Shaik F, Cristante E, Moseley TC, Madoo A, Rizzi M, Bainbridge JW, Tsoyi K, Rosas IO, Whiteford JR, De Rossi G. QM107, a novel CD148 (RTP Type J) activating peptide therapy for treating neovascular age-related macular degeneration. Br J Pharmacol 2025; 182:951-968. [PMID: 39428594 DOI: 10.1111/bph.17362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Angiogenesis is a pathological component of neovascular age-related macular degeneration. Current therapies, although successful, are prone to high levels of patient non-response and a loss of efficacy over time, indicating the need to explore other therapeutic avenues. We have shown that an interaction between syndecan-2 and the tyrosine phosphatase receptor CD148 (RTP Type J) results in the ablation of angiogenesis. Here we exploit this pathway to develop a peptide activator of CD148 as a therapy for neovascular age-related macular degeneration. EXPERIMENTAL APPROACH We tested a peptide (QM107) derived from syndecan-2 in a variety of angiogenesis models and a pre-clinical model of neovascular age-related macular degeneration. We assessed the toxicological and inflammatory profiles of QM107 and its stability in vitreous humour. KEY RESULTS QM107 inhibits angiogenesis in ex vivo sprouting assays and disrupts endothelial microcapillary formation via inhibition of cell migration. QM107 acts through CD148, leading to changes in GSK3A phosphorylation and β1 integrin activation. QM107 elicits a negligible inflammatory response and exhibits limited toxicity in cultured cells, and is stable in vitreous humour. Finally, we show proof of concept that QM107 blocks angiogenesis in vivo using a model of neovascular age-related macular degeneration. CONCLUSION AND IMPLICATIONS We have developed a CD148 activating peptide which shows promise in inhibiting angiogenesis in models of neovascular age-related macular degeneration. This treatment could either represent an alternative or augment existing therapies, and owing to its distinct mode of action be used in patients who do not respond to existing treatments.
Collapse
Affiliation(s)
- Samantha Arokiasamy
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michaela J M Balderstone
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Faheem Shaik
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Enrico Cristante
- Institute of Ophthalmology, University College London, London, UK
| | - Thomas C Moseley
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Akshay Madoo
- Institute of Ophthalmology, University College London, London, UK
| | - Matteo Rizzi
- Institute of Ophthalmology, University College London, London, UK
| | - James W Bainbridge
- Institute of Ophthalmology, University College London, London, UK
- NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Konstantin Tsoyi
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - James R Whiteford
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giulia De Rossi
- Institute of Ophthalmology, University College London, London, UK
| |
Collapse
|
10
|
Eldridge-Thomas BL, Bohere JG, Roubinet C, Barthelemy A, Samuels TJ, Teixeira FK, Kolahgar G. The transmembrane protein Syndecan is required for stem cell survival and maintenance of their nuclear properties. PLoS Genet 2025; 21:e1011586. [PMID: 39913561 PMCID: PMC11819509 DOI: 10.1371/journal.pgen.1011586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 02/12/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
Tissue maintenance is underpinned by resident stem cells whose activity is modulated by microenvironmental cues. Using Drosophila as a simple model to identify regulators of stem cell behaviour and survival in vivo, we have identified novel connections between the conserved transmembrane proteoglycan Syndecan, nuclear properties and stem cell function. In the Drosophila midgut, Syndecan depletion in intestinal stem cells results in their loss from the tissue, impairing tissue renewal. At the cellular level, Syndecan depletion alters cell and nuclear shape, and causes nuclear lamina invaginations and DNA damage. In a second tissue, the developing Drosophila brain, live imaging revealed that Syndecan depletion in neural stem cells results in nuclear envelope remodelling defects which arise upon cell division. Our findings reveal a new role for Syndecan in the maintenance of nuclear properties in diverse stem cell types.
Collapse
Affiliation(s)
- Buffy L. Eldridge-Thomas
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Jerome G. Bohere
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Chantal Roubinet
- Université de Rennes, CNRS, INSERM, IGDR (Institut de Génétique et Développement de Rennes), UMR 6290, ERL U1305, Rennes, France
| | - Alexandre Barthelemy
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Tamsin J. Samuels
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Felipe Karam Teixeira
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Golnar Kolahgar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Flores J, Nugent K. Sodium, the Vascular Endothelium, and Hypertension: A Narrative Review of Literature. Cardiol Rev 2025:00045415-990000000-00402. [PMID: 39807866 DOI: 10.1097/crd.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The vascular endothelium and its endothelial glycocalyx contribute to the protection of the endothelial cells from exposure to high levels of sodium and help these structures maintain normal function by regulating vascular permeability due to its buffering effect. The endothelial glycocalyx has negative surface charges that bind sodium and limit sodium entry into cells and the interstitial space. High sodium levels can disrupt this barrier and allow the movement of sodium into cells and extravascular fluid. This can generate reactive oxygen species that inhibit nitric oxide production. This leads to vasospasm and increases intravascular pressures. Overtime vascular remodeling occurs, and this changes the anatomy of blood vessels, their intrinsic stiffness, and their response to vasodilators and results in hypertension. Patients with increased salt sensitivity are potentially at more risk for this sequence of events. Studies on the degradation of the glycocalyx provide insight into the pathogenesis of clinical disorders with vascular involvement, but there is limited information available in the context of higher concentrations of sodium. Data on higher intake of sodium and the imbalance between nitric oxide and reactive oxygen species have been obtained in experimental studies and provide insights into possible outcomes in humans. The current western diet with sodium intake above recommended levels has led to the assessment of sodium sensitivity, which has been used in different populations and could become a practical tool to evaluate patients. This would potentially allow more focused recommendations regarding salt intake. This review will consider the structure of the vascular endothelium, its components, the effect of sodium on it, and the use of the salt blood test mini.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | |
Collapse
|
12
|
Lamba R, Paguntalan AM, Petrov PB, Naba A, Izzi V. MatriCom: a scRNA-Seq data mining tool to infer ECM-ECM and cell-ECM communication systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627834. [PMID: 39763937 PMCID: PMC11702561 DOI: 10.1101/2024.12.10.627834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
The ECM is a complex and dynamic meshwork of proteins that forms the framework of all multicellular organisms. Protein interactions within the ECM are critical to building and remodeling the ECM meshwork, while interactions between ECM proteins and cell surface receptors are essential for the initiation of signal transduction and the orchestration of cellular behaviors. Here, we report the development of MatriCom, a web application (https://matrinet.shinyapps.io/matricom) and a companion R package (https://github.com/Izzilab/MatriCom), devised to mine scRNA-Seq datasets and infer communications between ECM components and between different cell populations and the ECM. To impute interactions from expression data, MatriCom relies on a unique database, MatriComDB, that includes over 25,000 curated interactions involving matrisome components, with data on 80% of the ~1,000 genes that compose the mammalian matrisome. MatriCom offers the option to query open-access datasets sourced from large sequencing efforts (Tabula Sapiens, The Human Protein Atlas, HuBMAP) or to process user-generated datasets. MatriCom is also tailored to account for the specific rules governing ECM protein interactions and offers options to customize the output through stringency filters. We illustrate the usability of MatriCom with the example of the human kidney matrisome communication network. Last, we demonstrate how the integration of 46 scRNA-Seq datasets led to the identification of both ubiquitous and tissue-specific ECM communication patterns. We envision that MatriCom will become a powerful resource to elucidate the roles of different cell populations in ECM-ECM and cell-ECM interactions and their dysregulations in the context of diseases such as cancer or fibrosis.
Collapse
Affiliation(s)
- Rijuta Lamba
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
| | - Asia M. Paguntalan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Petar B. Petrov
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| |
Collapse
|
13
|
Nappi F, Nassif A, Schoell T. External Scaffold for Strengthening the Pulmonary Autograft in the Ross Procedure. Biomimetics (Basel) 2024; 9:674. [PMID: 39590246 PMCID: PMC11591583 DOI: 10.3390/biomimetics9110674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Despite offering several potential benefits over standard prosthetic aortic valve replacement, the use of the pulmonary autograft has been limited to date due to concerns over the risk of pulmonary autograft expansion and the need for reintervention. Several techniques using materials with biomimetic potential have been developed to reduce this complication. The incidence, risk factors, and pathophysiology of pulmonary autograft dilatation are discussed in this article. This seminar will provide an overview of the techniques of external pulmonary autograft support and their advantages and limitations. It also considers future directions for further investigation and future clinical applications of external pulmonary autograft support. Dilatation of the autograft is more likely to occur in patients with aortic regurgitation and a dilated aortic annulus. External scaffolding may prevent autograft stretching and expansion in these specific cases. However, from a biomimetic point of view, any permanent scaffold potentially restricts the movement of the autograft root. This reduces some of the benefits associated with the use of autologous tissue, which is the priority of the Ross procedure. To address this issue, several bioresorbable matrices could be used to support the root during its initial adaptive phase. Control of blood pressure with aggressive therapy is the first line to avoid this problem in the first year after pulmonary autograft implantation, together with support of the annular and sinotubular junction in some selected cases. This is the best way to maintain stable autograft root dimensions while preserving root dynamics. However, to determine the efficacy of this combined external support and best medical management, it is important to perform regular imaging and clinical follow-up.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, 93200 Saint-Denis, France; (A.N.); (T.S.)
| | | | | |
Collapse
|
14
|
Chen X, He F, Zhang H, Ma Y, Yu J, Qin H, Wu F, Wang Z, Zhan Y, Zhang J, Lu L, Zhang M, Yu S. Syndecan-4 inhibition attenuates cartilage degeneration in temporomandibular joint osteoarthritis. J Oral Rehabil 2024; 51:2324-2335. [PMID: 39101668 DOI: 10.1111/joor.13829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024]
Abstract
BACKGROUND Syndecan 4 (SDC4), a type I transmembrane proteoglycan, serves as a critical link between chondrocytes and the extracellular matrix. OBJECTIVE This study aimed to explore the role of SDC4 in cartilage degeneration of temporomandibular joint osteoathritis (TMJOA). METHODS Condylar chondrocytes were stimulated with varying concentrations of recombinant rat interleukin-1β (rrIL-1β) and SDC4 small interfering RNA (si-SDC4). Anti-SDC4 ectodomain-specific antibodies or IgG were intra-articularly administrated in a TMJOA model rats. SDC4 conditional knockout (SDC4-cKO) and Sdc4flox/flox mice were induced TMJOA. Cartilage degeneration was assessed using haematoxylin & eosin (H&E) and safranin O (SO) staining. Protein levels of SDC4, matrix metalloproteinases (MMPs), a disintegrin and metalloproteinase with a thrombospondin motifs 5 (ADAMTS5), tumour necrosis factor α (TNFα), type II collagen (Col-II), aggrecan (ACAN), cleaved caspase 3 (CASP3), Ki67 and related pathways in condylar cartilage were evaluated by immunohistochemical (IHC) staining or western blot assays. RESULTS SDC4 expression was evidently increased in MIA-model animals compared to control groups. rrIL-1β stimulation increased the expression of SDC4, MMP3 and ADAMTS5 expression in chondrocytes, while decreasing the expression of Col-II. These effects were reversed by si-SDC4 in vitro. In vivo, SDC4 blockade reduced the death of chondrocytes and the loss of cartilage matrix, which was evidenced by increased expression of Col-II and ACAN, and a decrease in SDC4, MMP13 and cleaved-CASP3-positive cells. Furthermore, the protein levels of ACAN and Ki67 were elevated, and the ERK1/2 and P38 signalling pathways were activated following SDC4 inhibition. CONCLUSIONS SDC4 inhibition significantly ameliorates condylar cartilage degeneration, which was mediated, at least partly, through P38 and ERK1/2 signalling. Inhibition of SDC4 may be of great value for the treatment of TMJOA.
Collapse
Affiliation(s)
- Xiaohua Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Feng He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Hongyun Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Yuanjun Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
- Department of Stomatology, Chinese PLA General Hospital of Central Theater Command, Wuhan, People's Republic of China
| | - Jia Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Han Qin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Zhuo Wang
- Lintong Xiekou Health Center, Xi'an, Shaanxi, People's Republic of China
| | - Ying Zhan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Lei Lu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Mian Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shibin Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Oral Anatomy and Physiology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
15
|
Lin PH, Xu Y, Bali SK, Kim J, Gimeno A, Roberts ET, James D, Almeida NMS, Loganathan N, Fan F, Wilson AK, Jonathan Amster I, Moremen KW, Liu J, Jiménez-Barbero J, Huang X. Solid-Phase-Supported Chemoenzymatic Synthesis and Analysis of Chondroitin Sulfate Proteoglycan Glycopeptides. Angew Chem Int Ed Engl 2024; 63:e202405671. [PMID: 38781001 PMCID: PMC11772155 DOI: 10.1002/anie.202405671] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Proteoglycans (PGs), consisting of glycosaminoglycans (GAGs) linked with the core protein through a tetrasaccharide linkage region, play roles in many important biological events. The chemical synthesis of PG glycopeptides is extremely challenging. In this work, the enzymes required for synthesis of chondroitin sulfate (CS) PG (CSPG) have been expressed and the suitable sequence of enzymatic reactions has been established. To expedite CSPG synthesis, the peptide acceptor was immobilized on solid phase and the glycan units were directly installed enzymatically onto the peptide. Subsequent enzymatic chain elongation and sulfation led to the successful synthesis of CSPG glycopeptides. The CS dodecasaccharide glycopeptide was the longest homogeneous CS glycopeptide synthesized to date. The enzymatic synthesis was much more efficient than the chemical synthesis of the corresponding CS glycopeptides, which could reduce the total number of synthetic steps by 80 %. The structures of the CS glycopeptides were confirmed by mass spectrometry analysis and NMR studies. In addition, the interactions between the CS glycopeptides and cathepsin G were studied. The sulfation of glycan chain was found to be important for binding with cathepsin G. This efficient chemoenzymatic strategy opens new avenues to investigate the structures and functions of PGs.
Collapse
Affiliation(s)
- Po-Han Lin
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, 27599, United States
| | - Semiha Kevser Bali
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Jandi Kim
- Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| | - Ana Gimeno
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CICbioGUNE), Basque Research and Technology Alliance (BRTA), 48160, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Elijah T Roberts
- Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| | - Deepak James
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Nuno M S Almeida
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Narasimhan Loganathan
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Fei Fan
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Angela K Wilson
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - I Jonathan Amster
- Department of Chemistry, University of Georgia, Athens, GA 30602, United States
| | - Kelley W Moremen
- Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, United States
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA 30602, United States
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, 27599, United States
| | - Jesús Jiménez-Barbero
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CICbioGUNE), Basque Research and Technology Alliance (BRTA), 48160, Derio, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48009, Spain
- Department of Inorganic & Organic Chemistry, Faculty of Science and Technology, University of the Basque Country, EHU-UPV, Leioa, 48940, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Madrid, 28029, Spain
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan, 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, 48824, United States
| |
Collapse
|
16
|
Ricard-Blum S, Vivès RR, Schaefer L, Götte M, Merline R, Passi A, Heldin P, Magalhães A, Reis CA, Skandalis SS, Karamanos NK, Perez S, Nikitovic D. A biological guide to glycosaminoglycans: current perspectives and pending questions. FEBS J 2024; 291:3331-3366. [PMID: 38500384 DOI: 10.1111/febs.17107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/08/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Mammalian glycosaminoglycans (GAGs), except hyaluronan (HA), are sulfated polysaccharides that are covalently attached to core proteins to form proteoglycans (PGs). This article summarizes key biological findings for the most widespread GAGs, namely HA, chondroitin sulfate/dermatan sulfate (CS/DS), keratan sulfate (KS), and heparan sulfate (HS). It focuses on the major processes that remain to be deciphered to get a comprehensive view of the mechanisms mediating GAG biological functions. They include the regulation of GAG biosynthesis and postsynthetic modifications in heparin (HP) and HS, the composition, heterogeneity, and function of the tetrasaccharide linkage region and its role in disease, the functional characterization of the new PGs recently identified by glycoproteomics, the selectivity of interactions mediated by GAG chains, the display of GAG chains and PGs at the cell surface and their impact on the availability and activity of soluble ligands, and on their move through the glycocalyx layer to reach their receptors, the human GAG profile in health and disease, the roles of GAGs and particular PGs (syndecans, decorin, and biglycan) involved in cancer, inflammation, and fibrosis, the possible use of GAGs and PGs as disease biomarkers, and the design of inhibitors targeting GAG biosynthetic enzymes and GAG-protein interactions to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Univ Lyon 1, ICBMS, UMR 5246 University Lyon 1 - CNRS, Villeurbanne cedex, France
| | | | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Germany
| | - Rosetta Merline
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | | | - Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Serge Perez
- Centre de Recherche sur les Macromolécules Végétales, University of Grenoble-Alpes, CNRS, France
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
17
|
Majchrzak K, Hentschel E, Hönzke K, Geithe C, von Maltzahn J. We need to talk-how muscle stem cells communicate. Front Cell Dev Biol 2024; 12:1378548. [PMID: 39050890 PMCID: PMC11266305 DOI: 10.3389/fcell.2024.1378548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Skeletal muscle is one of the tissues with the highest ability to regenerate, a finely controlled process which is critically depending on muscle stem cells. Muscle stem cell functionality depends on intrinsic signaling pathways and interaction with their immediate niche. Upon injury quiescent muscle stem cells get activated, proliferate and fuse to form new myofibers, a process involving the interaction of multiple cell types in regenerating skeletal muscle. Receptors in muscle stem cells receive the respective signals through direct cell-cell interaction, signaling via secreted factors or cell-matrix interactions thereby regulating responses of muscle stem cells to external stimuli. Here, we discuss how muscle stem cells interact with their immediate niche focusing on how this controls their quiescence, activation and self-renewal and how these processes are altered in age and disease.
Collapse
Affiliation(s)
- Karolina Majchrzak
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Erik Hentschel
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Katja Hönzke
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Christiane Geithe
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
| | - Julia von Maltzahn
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus–Senftenberg, Senftenberg, Germany
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
- Faculty for Environment and Natural Sciences, Brandenburg University of Technology Cottbus—Senftenberg, Senftenberg, Germany
| |
Collapse
|
18
|
Roncali L, Marionneau-Lambot S, Roy C, Eychenne R, Gouard S, Avril S, Chouin N, Riou J, Allard M, Rousseau A, Guérard F, Hindré F, Chérel M, Garcion E. Brain intratumoural astatine-211 radiotherapy targeting syndecan-1 leads to durable glioblastoma remission and immune memory in female mice. EBioMedicine 2024; 105:105202. [PMID: 38905749 PMCID: PMC11246004 DOI: 10.1016/j.ebiom.2024.105202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Glioblastoma (GB), the most aggressive brain cancer, remains a critical clinical challenge due to its resistance to conventional treatments. Here, we introduce a locoregional targeted-α-therapy (TAT) with the rat monoclonal antibody 9E7.4 targeting murine syndecan-1 (SDC1) coupled to the α-emitter radionuclide astatine-211 (211At-9E7.4). METHODS We orthotopically transplanted 50,000 GL261 cells of murine GB into the right striatum of syngeneic female C57BL/6JRj mice using stereotaxis. After MRI validation of tumour presence at day 11, TAT was injected at the same coordinates. Biodistribution, efficacy, toxicity, local and systemic responses were assessed following application of this protocol. The 9E7.4 monoclonal antibody was labelled with iodine-125 (125I) for biodistribution and with astatine-211 (211At) for the other experiments. FINDINGS The 211At-9E7.4 TAT demonstrated robust efficacy in reducing orthotopic tumours and achieved improved survival rates in the C57BL/6JRj model, reaching up to 70% with a minimal activity of 100 kBq. Targeting SDC1 ensured the cerebral retention of 211At over an optimal time window, enabling low-activity administration with a minimal toxicity profile. Moreover, TAT substantially reduced the occurrence of secondary tumours and provided resistance to new tumour development after contralateral rechallenge, mediated through the activation of central and effector memory T cells. INTERPRETATION The locoregional 211At-9E7.4 TAT stands as one of the most efficient TAT across all preclinical GB models. This study validates SDC1 as a pertinent therapeutic target for GB and underscores 211At-9E7.4 TAT as a promising advancement to improve the treatment and quality of life for patients with GB. FUNDING This work was funded by the French National Agency for Research (ANR) "France 2030 Investment Plan" Labex Iron [ANR-11-LABX-18-01], The SIRIC ILIAD [INCa-DGOS-INSERM-18011], the French program "Infrastructure d'Avenir en Biologie-Santé" (France Life Imaging) [ANR-11-INBS-0006], the PIA3 of the ANR, integrated to the "France 2030 Investment Plan" [ANR-21-RHUS-0012], and support from Inviscan SAS (Strasbourg, France). It was also related to: the ANR under the frame of EuroNanoMed III (project GLIOSILK) [ANR-19-ENM3-0003-01]; the "Région Pays-de-la-Loire" under the frame of the Target'In project; the "Ligue Nationale contre le Cancer" and the "Comité Départemental de Maine-et-Loire de la Ligue contre le Cancer" (CD49) under the frame of the FusTarG project and the "Tumour targeting, imaging and radio-therapies network" of the "Cancéropôle Grand-Ouest" (France). This work was also funded by the Institut National de la Santé et de la Recherche Médicale (INSERM), the University of Nantes, and the University of Angers.
Collapse
Affiliation(s)
- Loris Roncali
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Séverine Marionneau-Lambot
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CHU Nantes, Nantes Université, Service de médecine nucléaire, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Charlotte Roy
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Romain Eychenne
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; GIP ARRONAX, F-44160, Saint-Herblain, France
| | - Sébastien Gouard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Sylvie Avril
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France
| | - Nicolas Chouin
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; ONIRIS, F-44000, Nantes, France
| | - Jérémie Riou
- CHU Angers, Université d'Angers, F-49000, Angers, France
| | - Mathilde Allard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Audrey Rousseau
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; CHU Angers, Université d'Angers, F-49000, Angers, France
| | - François Guérard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - François Hindré
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Michel Chérel
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; Institut de Cancérologie de l'Ouest, Service de médecine nucléaire, F-44160, Saint-Herblain, France.
| | - Emmanuel Garcion
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France; PACEM (Plateforme d'Analyse Cellulaire et Moléculaire), Université d'Angers, SFR 4208, F-49000, Angers, France.
| |
Collapse
|
19
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
20
|
Sao K, Risbud MV. Proteoglycan Dysfunction: A Common Link Between Intervertebral Disc Degeneration and Skeletal Dysplasia. Neurospine 2024; 21:162-178. [PMID: 38569642 PMCID: PMC10992626 DOI: 10.14245/ns.2347342.671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/04/2024] [Accepted: 02/23/2024] [Indexed: 04/05/2024] Open
Abstract
Proteoglycans through their sulfated glycosaminoglycans regulate cell-matrix signaling during tissue development, regeneration, and degeneration processes. Large extracellular proteoglycans such as aggrecan, versican, and perlecan are especially important for the structural integrity of the intervertebral disc and cartilage during development. In these tissues, proteoglycans are responsible for hydration, joint flexibility, and the absorption of mechanical loads. Loss or reduction of these molecules can lead to disc degeneration and skeletal dysplasia, evident from loss of disc height or defects in skeletal development respectively. In this review, we discuss the common proteoglycans found in the disc and cartilage and elaborate on various murine models and skeletal dysplasias in humans to highlight how their absence and/or aberrant expression causes accelerated disc degeneration and developmental defects.
Collapse
Affiliation(s)
- Kimheak Sao
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V. Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
21
|
Yang L, Sheets TP, Feng Y, Yu G, Bajgain P, Hsu KS, So D, Seaman S, Lee J, Lin L, Evans CN, Guest MR, Chari R, St. Croix B. Uncovering receptor-ligand interactions using a high-avidity CRISPR activation screening platform. SCIENCE ADVANCES 2024; 10:eadj2445. [PMID: 38354234 PMCID: PMC10866537 DOI: 10.1126/sciadv.adj2445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
The majority of clinically approved drugs target proteins that are secreted or cell surface bound. However, further advances in this area have been hindered by the challenging nature of receptor deorphanization, as there are still many secreted and cell-bound proteins with unknown binding partners. Here, we developed an advanced screening platform that combines CRISPR-CAS9 guide-mediated gene activation (CRISPRa) and high-avidity bead-based selection. The CRISPRa platform incorporates serial enrichment and flow cytometry-based monitoring, resulting in substantially improved screening sensitivity for well-known yet weak interactions of the checkpoint inhibitor family. Our approach has successfully revealed that siglec-4 exerts regulatory control over T cell activation through a low affinity trans-interaction with the costimulatory receptor 4-1BB. Our highly efficient screening platform holds great promise for identifying extracellular interactions of uncharacterized receptor-ligand partners, which is essential to develop next-generation therapeutics, including additional immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Liping Yang
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Timothy P. Sheets
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21702, USA
| | - Yang Feng
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Guojun Yu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Pradip Bajgain
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Kuo-Sheng Hsu
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Daeho So
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Steven Seaman
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Jaewon Lee
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| | - Ling Lin
- Proteomic Instability of Cancer Section, MCGP, NCI, NIH, Frederick, MD 21702, USA
| | - Christine N. Evans
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21702, USA
| | - Mary R. Guest
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21702, USA
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Lab for Cancer Research, Frederick, MD 21702, USA
| | - Brad St. Croix
- Tumor Angiogenesis Unit, Mouse Cancer Genetics Program (MCGP), National Cancer Institute (NCI), NIH, Frederick, MD 21702, USA
| |
Collapse
|
22
|
Kim M, Kim Y. NMR Structural Study of Syndecan-4 Transmembrane Domain with Cytoplasmic Region. Molecules 2023; 28:7855. [PMID: 38067582 PMCID: PMC10708377 DOI: 10.3390/molecules28237855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/27/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Syndecan-4 (SDC4) consists of transmembrane heparan sulfate proteoglycan (HSPG) belonging to the syndecan family. It is present in most cell types of Mammalia. Its structure contains a heparan-sulfate-modified extracellular domain, a single transmembrane domain, and a short C-terminal cytoplasmic domain. Regarding the overall cellular function of SDC4, other cells or ligands can bind to its ecto-domain. In addition, 4,5-bisphosphate phosphatidylinositol (PIP2) or protein kinase Cα can bind to its cyto-domain to activate downstream signaling pathways. To understand the signal transduction mechanism of syndecan, it is important to know the interactions between their actual structure and function in vivo. Therefore, it is important to identify the structure of SDC4 to understand the ligand binding behavior of SDC4. In this study, expression and purification were performed to reveal structures of the short ecto-domain, the transmembrane domain, and the cytoplasmic domain of Syd4-eTC (SDC4). Solution-state NMR spectroscopy and solid-state NMR spectroscopy were used to study the structure of Syd4-eTC in membrane environments and to demonstrate the interaction between Syd4-eTC and PIP2.
Collapse
Affiliation(s)
| | - Yongae Kim
- Department of Chemistry, Hankuk University of Foreign Studies, 81 Oedae-ro, Mohyeon, Yongin 17035, Republic of Korea;
| |
Collapse
|
23
|
Novoseletskaya ES, Evdokimov PV, Efimenko AY. Extracellular matrix-induced signaling pathways in mesenchymal stem/stromal cells. Cell Commun Signal 2023; 21:244. [PMID: 37726815 PMCID: PMC10507829 DOI: 10.1186/s12964-023-01252-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/31/2023] [Indexed: 09/21/2023] Open
Abstract
The extracellular matrix (ECM) is a crucial component of the stem cell microenvironment, or stem-cell niches, and contributes to the regulation of cell behavior and fate. Accumulating evidence indicates that different types of stem cells possess a large variety of molecules responsible for interactions with the ECM, mediating specific epigenetic rearrangements and corresponding changes in transcriptome profile. Signals from the ECM are crucial at all stages of ontogenesis, including embryonic and postnatal development, as well as tissue renewal and repair. The ECM could regulate stem cell transition from a quiescent state to readiness to perceive the signals of differentiation induction (competence) and the transition between different stages of differentiation (commitment). Currently, to unveil the complex networks of cellular signaling from the ECM, multiple approaches including screening methods, the analysis of the cell matrixome, and the creation of predictive networks of protein-protein interactions based on experimental data are used. In this review, we consider the existing evidence regarded the contribution of ECM-induced intracellular signaling pathways into the regulation of stem cell differentiation focusing on mesenchymal stem/stromal cells (MSCs) as well-studied type of postnatal stem cells totally depended on signals from ECM. Furthermore, we propose a system biology-based approach for the prediction of ECM-mediated signal transduction pathways in target cells. Video Abstract.
Collapse
Affiliation(s)
- Ekaterina Sergeevna Novoseletskaya
- Faculty of Biology, Dayun New Town, Shenzhen MSU-BIT University, 1 International University Park Road, Dayun New Town, Longgang District, Shenzhen, Guangdong Province, P. R. China.
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosov Ave., 27/10, 119991, Moscow, Russia.
| | - Pavel Vladimirovich Evdokimov
- Materials Science Department, Lomonosov Moscow State University, Leninskie Gory, 1, Building 73, 119991, Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, GSP-1, Leninskiye Gory, 1-3, Moscow, Russia
| | - Anastasia Yurievna Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosov Ave., 27/10, 119991, Moscow, Russia
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosov Ave., 27/1, 119991, Moscow, Russia
| |
Collapse
|
24
|
Xu Z, Chen Y, Wang Y, Han W, Xu W, Liao X, Zhang T, Wang G. Matrix stiffness, endothelial dysfunction and atherosclerosis. Mol Biol Rep 2023; 50:7027-7041. [PMID: 37382775 DOI: 10.1007/s11033-023-08502-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 04/28/2023] [Indexed: 06/30/2023]
Abstract
Atherosclerosis (AS) is the leading cause of the human cardiovascular diseases (CVDs). Endothelial dysfunction promotes the monocytes infiltration and inflammation that participate fundamentally in atherogenesis. Endothelial cells (EC) have been recognized as mechanosensitive cells and have different responses to distinct mechanical stimuli. Emerging evidence shows matrix stiffness-mediated EC dysfunction plays a vital role in vascular disease, but the underlying mechanisms are not yet completely understood. This article aims to summarize the effect of matrix stiffness on the pro-atherosclerotic characteristics of EC including morphology, rigidity, biological behavior and function as well as the related mechanical signal. The review also discusses and compares the contribution of matrix stiffness-mediated phagocytosis of macrophages and EC to AS progression. These advances in our understanding of the relationship between matrix stiffness and EC dysfunction open the avenues to improve the prevention and treatment of now-ubiquitous atherosclerotic diseases.
Collapse
Affiliation(s)
- Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Yi Chen
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Wenbo Han
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Wenfeng Xu
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Xiaoling Liao
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Tao Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection, Chongqing Key Laboratory of Nano/Micro Composite Material and Device, School of Metallurgy and Materials Engineering, Chongqing University of Science and Technology, Chongqing, 401331, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400030, China.
- Bioengineering College of Chongqing University, NO.174, Shazheng Street, Shapingba District, Chongqing, 400030, PR China.
| |
Collapse
|
25
|
Sembajwe LF, Ssekandi AM, Namaganda A, Muwonge H, Kasolo JN, Kalyesubula R, Nakimuli A, Naome M, Patel KP, Masenga SK, Kirabo A. Glycocalyx-Sodium Interaction in Vascular Endothelium. Nutrients 2023; 15:2873. [PMID: 37447199 PMCID: PMC10343370 DOI: 10.3390/nu15132873] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
The glycocalyx generally covers almost all cellular surfaces, where it participates in mediating cell-surface interactions with the extracellular matrix as well as with intracellular signaling molecules. The endothelial glycocalyx that covers the luminal surface mediates the interactions of endothelial cells with materials flowing in the circulating blood, including blood cells. Cardiovascular diseases (CVD) remain a major cause of morbidity and mortality around the world. The cardiovascular risk factors start by causing endothelial cell dysfunction associated with destruction or irregular maintenance of the glycocalyx, which may culminate into a full-blown cardiovascular disease. The endothelial glycocalyx plays a crucial role in shielding the cell from excessive exposure and absorption of excessive salt, which can potentially cause damage to the endothelial cells and underlying tissues of the blood vessels. So, in this mini review/commentary, we delineate and provide a concise summary of the various components of the glycocalyx, their interaction with salt, and subsequent involvement in the cardiovascular disease process. We also highlight the major components of the glycocalyx that could be used as disease biomarkers or as drug targets in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lawrence Fred Sembajwe
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Abdul M. Ssekandi
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Agnes Namaganda
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Haruna Muwonge
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Josephine N. Kasolo
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Robert Kalyesubula
- Department of Medical Physiology, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda; (A.M.S.); (A.N.); (H.M.); (J.N.K.); (R.K.)
| | - Annettee Nakimuli
- Department of Obstetrics and Gynecology, School of Medicine, Makerere University College of Health Sciences, Kampala P.O. Box 7072, Uganda;
| | - Mwesigwa Naome
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Sepiso K. Masenga
- Department of Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Kabwe P.O. Box 80415, Zambia;
| | - Annet Kirabo
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
26
|
Ricard-Blum S, Couchman JR. Conformations, interactions and functions of intrinsically disordered syndecans. Biochem Soc Trans 2023:BST20221085. [PMID: 37334846 DOI: 10.1042/bst20221085] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans present on most mammalian cell surfaces. They have a long evolutionary history, a single syndecan gene being expressed in bilaterian invertebrates. Syndecans have attracted interest because of their potential roles in development and disease, including vascular diseases, inflammation and various cancers. Recent structural data is providing important insights into their functions, which are complex, involving both intrinsic signaling through cytoplasmic binding partners and co-operative mechanisms where syndecans form a signaling nexus with other receptors such as integrins and tyrosine kinase growth factor receptors. While the cytoplasmic domain of syndecan-4 has a well-defined dimeric structure, the syndecan ectodomains are intrinsically disordered, which is linked to a capacity to interact with multiple partners. However, it remains to fully establish the impact of glycanation and partner proteins on syndecan core protein conformations. Genetic models indicate that a conserved property of syndecans links the cytoskeleton to calcium channels of the transient receptor potential class, compatible with roles as mechanosensors. In turn, syndecans influence actin cytoskeleton organization to impact motility, adhesion and the extracellular matrix environment. Syndecan clustering with other cell surface receptors into signaling microdomains has relevance to tissue differentiation in development, for example in stem cells, but also in disease where syndecan expression can be markedly up-regulated. Since syndecans have potential as diagnostic and prognostic markers as well as possible targets in some forms of cancer, it remains important to unravel structure/function relationships in the four mammalian syndecans.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- ICBMS, UMR 5246 CNRS, Universite Claude Bernard Lyon 1, F-69622 Villeurbanne, France
| | - John R Couchman
- Biotech Research & Innovation Center, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
27
|
Sztretye M, Singlár Z, Ganbat N, Al-Gaadi D, Szabó K, Köhler ZM, Dux L, Keller-Pintér A, Csernoch L, Szentesi P. Unravelling the Effects of Syndecan-4 Knockdown on Skeletal Muscle Functions. Int J Mol Sci 2023; 24:ijms24086933. [PMID: 37108098 PMCID: PMC10138797 DOI: 10.3390/ijms24086933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The remodelling of the extracellular matrix plays an important role in skeletal muscle development and regeneration. Syndecan-4 is a cell surface proteoglycan crucial for muscle differentiation. Syndecan-4-/- mice have been reported to be unable to regenerate following muscle damage. To investigate the consequences of the decreased expression of Syndecan-4, we have studied the in vivo and in vitro muscle performance and the excitation-contraction coupling machinery in young and aged Syndecan-4+/- (SDC4) mice. In vivo grip force was decreased significantly as well as the average and maximal speed of voluntary running in SDC4 mice, regardless of their age. The maximal in vitro twitch force was reduced in both EDL and soleus muscles from young and aged SDC4 mice. Ca2+ release from the sarcoplasmic reticulum decreased significantly in the FDB fibres of young SDC4 mice, while its voltage dependence was unchanged regardless of age. These findings were present in muscles from young and aged mice as well. On C2C12 murine skeletal muscle cells, we have also found altered calcium homeostasis upon Syndecan-4 silencing. The decreased expression of Syndecan-4 leads to reduced skeletal muscle performance in mice and altered motility in C2C12 myoblasts via altered calcium homeostasis. The altered muscle force performance develops at an early age and is maintained throughout the life course of the animal until old age.
Collapse
Affiliation(s)
- Mónika Sztretye
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, 4032 Debrecen, Hungary
| | - Zoltán Singlár
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Nyamkhuu Ganbat
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Dána Al-Gaadi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Kitti Szabó
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Márton Köhler
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - László Dux
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Anikó Keller-Pintér
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- ELKH-DE Cell Physiology Research Group, 4032 Debrecen, Hungary
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
28
|
Revisiting the Syndecans: Master Signaling Regulators with Prognostic and Targetable Therapeutic Values in Breast Carcinoma. Cancers (Basel) 2023; 15:cancers15061794. [PMID: 36980680 PMCID: PMC10046401 DOI: 10.3390/cancers15061794] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Syndecans (SDC1 to 4), a family of cell surface heparan sulfate proteoglycans, are frequently expressed in mammalian tissues. SDCs are aberrantly expressed either on tumor or stromal cells, influencing cancer initiation and progression through their pleiotropic role in different signaling pathways relevant to proliferation, cell-matrix adhesion, migration, invasion, metastasis, cancer stemness, and angiogenesis. In this review, we discuss the key roles of SDCs in the pathogenesis of breast cancer, the most common malignancy in females worldwide, focusing on the prognostic significance and molecular regulators of SDC expression and localization in either breast tumor tissue or its microenvironmental cells and the SDC-dependent epithelial–mesenchymal transition program. This review also highlights the molecular mechanisms underlying the roles of SDCs in regulating breast cancer cell behavior via modulation of nuclear hormone receptor signaling, microRNA expression, and exosome biogenesis and functions, as well as summarizing the potential of SDCs as promising candidate targets for therapeutic strategies against breast cancer.
Collapse
|
29
|
Colin-Pierre C, El Baraka O, Danoux L, Bardey V, André V, Ramont L, Brézillon S. Regulation of stem cell fate by HSPGs: implication in hair follicle cycling. NPJ Regen Med 2022; 7:77. [PMID: 36577752 PMCID: PMC9797564 DOI: 10.1038/s41536-022-00267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/30/2022] [Indexed: 12/29/2022] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) are part of proteoglycan family. They are composed of heparan sulfate (HS)-type glycosaminoglycan (GAG) chains covalently linked to a core protein. By interacting with growth factors and/or receptors, they regulate numerous pathways including Wnt, hedgehog (Hh), bone morphogenic protein (BMP) and fibroblast growth factor (FGF) pathways. They act as inhibitor or activator of these pathways to modulate embryonic and adult stem cell fate during organ morphogenesis, regeneration and homeostasis. This review summarizes the knowledge on HSPG structure and classification and explores several signaling pathways regulated by HSPGs in stem cell fate. A specific focus on hair follicle stem cell fate and the possibility to target HSPGs in order to tackle hair loss are discussed in more dermatological and cosmeceutical perspectives.
Collapse
Affiliation(s)
- Charlie Colin-Pierre
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France.
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France.
- BASF Beauty Care Solutions France SAS, Pulnoy, France.
| | | | - Louis Danoux
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | | | - Valérie André
- BASF Beauty Care Solutions France SAS, Pulnoy, France
| | - Laurent Ramont
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
- CHU de Reims, Service Biochimie-Pharmacologie-Toxicologie, Reims, France
| | - Stéphane Brézillon
- Université de Reims Champagne-Ardenne, SFR CAP-Santé (FED 4231), Laboratoire de Biochimie Médicale et Biologie Moléculaire, Reims, France
- CNRS UMR 7369, Matrice Extracellulaire et Dynamique Cellulaire-MEDyC, Reims, France
| |
Collapse
|
30
|
Takahashi I. Importance of Heparan Sulfate Proteoglycans in Pancreatic Islets and β-Cells. Int J Mol Sci 2022; 23:12082. [PMID: 36292936 PMCID: PMC9603760 DOI: 10.3390/ijms232012082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
β-cells in the islets of Langerhans of the pancreas secrete insulin in response to the glucose concentration in the blood. When these pancreatic β-cells are damaged, diabetes develops through glucose intolerance caused by insufficient insulin secretion. High molecular weight polysaccharides, such as heparin and heparan sulfate (HS) proteoglycans, and HS-degrading enzymes, such as heparinase, participate in the protection, maintenance, and enhancement of the functions of pancreatic islets and β-cells, and the demand for studies on glycobiology within the field of diabetes research has increased. This review introduces the roles of complex glycoconjugates containing high molecular weight polysaccharides and their degrading enzymes in pancreatic islets and β-cells, including those obtained in studies conducted by us earlier. In addition, from the perspective of glycobiology, this study proposes the possibility of application to diabetes medicine.
Collapse
Affiliation(s)
- Iwao Takahashi
- Division of Molecular and Cellular Pharmacology, Department of Pathophysiology and Pharmacology, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Morioka 028-3694, Iwate, Japan
| |
Collapse
|
31
|
Anderson AR, Cook GA. Recombinant expression, purification, and structural analysis of two ectodomains of Syndecan-1. Protein Expr Purif 2022; 201:106170. [PMID: 36179942 DOI: 10.1016/j.pep.2022.106170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022]
Abstract
Syndecan-1 (SDC-1) is an integral membrane heparin sulfate proteoglycan that is involved in inflammatory response, cell-signaling, cell proliferation, and numerous other cell-matrix interactions. Like the other members of the syndecan family, very little is known about structural conformations and dynamics of SDC-1. A majority of interactions occur through the extracellular ectodomain, therefore we have dedicated our research efforts to the study this specific portion of SDC-1. The ectodomain is often shed from the cell surface due to various stimuli. The released fragment has already been used as a useful biomarker for prognosis of some diseases and cancers. SDC-1 can be cleaved in different locations depending on the sheddase, generating soluble shed ectodomains that can be carried away in blood sera. In this study, we focus specifically on two main cleavage fragments that can be generated. We show the first successful expression and purification of recombinant SDC-1 ectodomains. Production of SDC-1 in E. coli allows the production of the core protein without risking heterogeneous post-translational modifications such as glycosylation, allowing a certain level of control over protein homogeneity that is not possible in mammalian expression. An expression vector was used to generate two different fusion proteins consisting of a His-tag and a TEV cleavage site for the removal of the fusion partner. SDS-PAGE was used to track the expression as well as the purification. Masses of the isolated proteins were determined using mass spectrometry and the purity and homogeneity were evaluated by solution NMR.
Collapse
Affiliation(s)
- Austin R Anderson
- Department of Chemistry, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Gabriel A Cook
- Department of Chemistry, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
32
|
Jia X, Zhu Z, Miao J, Zhang L, Li X, Bao Y, Jiang M. Serum Syndecan-1 levels in patients with immunoglobulin A vasculitis in children. J Pediatr (Rio J) 2022; 98:526-532. [PMID: 35240047 PMCID: PMC9510791 DOI: 10.1016/j.jped.2022.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the serum Syndecan-1 (SDC-1) levels in patients with immunoglobulin-A vasculitis (IgAV) in children and its relation with gastrointestinal involvements. METHODS Sixty-eight children with IgAV and 48 healthy children were enrolled in this cross-sectional study. Clinical and related laboratory data were collected from a computerized hospital database. Serum SDC-1 was collected on admission prior to treatment. RESULTS Forty-eight patients fully met the IgAV diagnostic criteria at admission (IgAV group), 20 patients with rash only and diagnosed IgAV during hospitalization (Purpura group). In IgAV group, 30 patients with gastrointestinal involvements (IgAV-GI group) and 18 patients without gastrointestinal involvements (IgAV-NGI group). SDC-1 serum levels were significantly higher in the IgAV group (86.37 ng/mL (IQR 59.16-117.14 ng/mL)) than in the controls (20.37 ng/mL (IQR 15.52-26.45 ng/mL)) and the Purpura group (32.66 ng/mL (IQR 14.87-49.89 ng/mL)). Additionally, SDC-1 (OR = 1.08) was independently associated with IgAV with a cut-off value (sensitivity and specificity) of 66.55 ng/mL (68.8%, 95.0%), and the area under the curve was 0.908. The serum SDC-1 levels of the IgAV-GI group (106.92 ± 50.12 ng/mL) were significantly higher than those in the IgAV-NGI group (67.52 ± 17.59 ng/mL). Logistic regression analysis showed that SDC-1 (OR = 1.03) was independently associated with IgAV-GI with a cut-off value of 89.39 ng/mL. CONCLUSIONS SDC-1 serum levels may mirror vascular endothelium injury and mucosal damage in IgAV. Its applicability as a surrogate biomarker in IgAV remains to be determined.
Collapse
Affiliation(s)
- Xinyi Jia
- Gastrointestinal Lab, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China; Jinhua Hospital, Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China; Endoscopy center and Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Zhenya Zhu
- Gastrointestinal Lab, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China
| | - Jing Miao
- Gastrointestinal Lab, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China; Jinhua Hospital, Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Linqian Zhang
- Jinhua Hospital, Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Xiaobing Li
- Jinhua Hospital, Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Yunguang Bao
- Jinhua Hospital, Zhejiang University and Jinhua Municipal Central Hospital, Jinhua, China
| | - Mizu Jiang
- Gastrointestinal Lab, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China; Endoscopy center and Department of Gastroenterology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Hangzhou, Zhejiang, China.
| |
Collapse
|
33
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
34
|
Critcher M, Huang ML. Excavating proteoglycan structure-function relationships: Modern approaches to capture the interactions of ancient biomolecules. Am J Physiol Cell Physiol 2022; 323:C415-C422. [PMID: 35759439 PMCID: PMC9359657 DOI: 10.1152/ajpcell.00222.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteoglycans are now well regarded as key facilitators of cell biology. While a majority of their interactions and functions are attributed to the decorating glycosaminoglycan chains, there is a growing appreciation for the roles of the proteoglycan core protein and for considering proteoglycans as replete protein-glycan conjugates. This appreciation, seeded by early work in proteoglycan biology, is now being advanced and exalted by modern approaches in chemical glycobiology. In this review, we discuss up-and-coming methods to unearth the fine-scale architecture of proteoglycans that modulate their functions and interactions. Crucial to these efforts is the production of chemically defined materials, including semi-synthetic proteoglycans and the in situ capture of interacting proteins. Together, the integration of chemical biology approaches promises to expedite the dissection of the structural heterogeneity of proteoglycans and deliver refined insight into their functions.
Collapse
Affiliation(s)
- Meg Critcher
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, La Jolla, CA.,Department of Molecular Medicine, Scripps Research, La Jolla, CA
| | - Mia L Huang
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, La Jolla, CA.,Department of Molecular Medicine, Scripps Research, La Jolla, CA.,Department of Chemistry, Scripps Research, La Jolla, CA
| |
Collapse
|
35
|
Glycosaminoglycan interaction networks and databases. Curr Opin Struct Biol 2022; 74:102355. [DOI: 10.1016/j.sbi.2022.102355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/14/2022]
|
36
|
The Cell Surface Heparan Sulfate Proteoglycan Syndecan-3 Promotes Ovarian Cancer Pathogenesis. Int J Mol Sci 2022; 23:ijms23105793. [PMID: 35628603 PMCID: PMC9145288 DOI: 10.3390/ijms23105793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 11/24/2022] Open
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans that integrate signaling at the cell surface. By interacting with cytokines, signaling receptors, proteases, and extracellular matrix proteins, syndecans regulate cell proliferation, metastasis, angiogenesis, and inflammation. We analyzed public gene expression datasets to evaluate the dysregulation and potential prognostic impact of Syndecan-3 in ovarian cancer. Moreover, we performed functional in vitro analysis in syndecan-3-siRNA-treated SKOV3 and CAOV3 ovarian cancer cells. In silico analysis of public gene array datasets revealed that syndecan-3 mRNA expression was significantly increased 5.8-fold in ovarian cancer tissues (n = 744) and 3.4-fold in metastases (n = 44) compared with control tissue (n = 46), as independently confirmed in an RNAseq dataset on ovarian serous cystadenocarcinoma tissue (n = 374, controls: n = 133, 3.5-fold increase tumor vs. normal). Syndecan-3 siRNA knockdown impaired 3D spheroid growth and colony formation as stemness-related readouts in SKOV3 and CAOV3 cells. In SKOV3, but not in CAOV3 cells, syndecan-3 depletion reduced cell viability both under basal conditions and under chemotherapy with cisplatin, or cisplatin and paclitaxel. While analysis of the SIOVDB database did not reveal differences in Syndecan-3 expression between patients, sensitive, resistant or refractory to chemotherapy, KM Plotter analysis of 1435 ovarian cancer patients revealed that high syndecan-3 expression was associated with reduced survival in patients treated with taxol and platin. At the molecular level, a reduction in Stat3 activation and changes in the expression of Wnt and notch signaling constituents were observed. Our study suggests that up-regulation of syndecan-3 promotes the pathogenesis of ovarian cancer by modulating stemness-associated pathways.
Collapse
|
37
|
Vallet SD, Berthollier C, Ricard-Blum S. The glycosaminoglycan interactome 2.0. Am J Physiol Cell Physiol 2022; 322:C1271-C1278. [PMID: 35544698 DOI: 10.1152/ajpcell.00095.2022] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glycosaminoglycans (GAGs) are complex linear polysaccharides, which are covalently attached to core proteins (except for hyaluronan) to form proteoglycans. They play key roles in the organization of the extracellular matrix, and at the cell surface where they contribute to the regulation of cell signaling and of cell adhesion. To explore the mechanisms and pathways underlying their functions, we have generated an expanded dataset of 4290 interactions corresponding to 3464 unique GAG-binding proteins, four times more than the first version of the GAG interactome (Vallet and Ricard-Blum, 2021 J Histochem Cytochem 69:93-104). The increased size of the GAG network is mostly due to the addition of GAG-binding proteins captured from cell lysates and biological fluids by affinity chromatography and identified by mass spectrometry. We review here the interaction repertoire of natural GAGs and of synthetic sulfated hyaluronan, the specificity and molecular functions of GAG-binding proteins, and the biological processes and pathways they are involved in. This dataset is also used to investigate the differences between proteins binding to iduronic acid-containing GAGs (dermatan sulfate and heparin/heparan sulfate) and those interacting with GAGs lacking iduronic acid (chondroitin sulfate, hyaluronan, and keratan sulfate).
Collapse
|
38
|
Kontio J, Soñora VR, Pesola V, Lamba R, Dittmann A, Navarro AD, Koivunen J, Pihlajaniemi T, Izzi V. Analysis of extracellular matrix network dynamics in cancer using the MatriNet database. Matrix Biol 2022; 110:141-150. [DOI: 10.1016/j.matbio.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/23/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
|
39
|
Shaik F, Balderstone MJM, Arokiasamy S, Whiteford JR. Roles of Syndecan-4 in cardiac injury and repair. Int J Biochem Cell Biol 2022; 146:106196. [PMID: 35331918 DOI: 10.1016/j.biocel.2022.106196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
The heparan sulphate proteoglycan Syndecan-4 belongs to a 4-member family of transmembrane receptors. Genetic deletion of Syndecan-4 in mice causes negligible developmental abnormalities however when challenged these animals show distinct phenotypes. Synedcan-4 is expressed in many cell types in the heart and its expression is elevated in response to cardiac injury and recent studies have suggested roles for Syndecan-4 in repair mechanisms within the damaged heart. The purpose of this review is to explore these biological insights into the role of Syndecan-4 in both the injured heart and later during cardiac repair and remodeling.
Collapse
Affiliation(s)
- Faheem Shaik
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK
| | - Michaela J M Balderstone
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK
| | - Samantha Arokiasamy
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK.
| | - James R Whiteford
- William Harvey Research Institute, Centre for Microvascular Research, Faculty of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, EC1M 6BQ, UK.
| |
Collapse
|
40
|
Fujikawa K, Nonaka N, Wang X, Shibata S. An in situ hybridization study of syndecan family during the late stages of developing mouse molar tooth germ. Anat Sci Int 2022; 97:358-368. [PMID: 35119611 DOI: 10.1007/s12565-022-00647-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/01/2022] [Indexed: 11/27/2022]
Abstract
Expression of syndecan-1, 2, 3, and 4 mRNAs during the late stages of tooth germ formation was investigated by in situ hybridization, using [35S]-UTP-labeled cRNA probes. Syndecan-1 mRNA was mainly expressed in the stellate reticulum and stratum intermedium as well as at the cervical region of dental papilla/dental follicle during E18.5-P3.0. Expression in the dental epithelium was enhanced during the postnatal periods, which was supported by real-time RT-PCR analysis. These spatiotemporal expression patterns may suggest specific roles of syndecan-1 in tooth formation such as tooth eruption or root formation. Syndecan-3 mRNA expression became evident in odontoblasts at E18.5, but compared to collagen type I mRNA, which was strongly expressed at this stage, syndecan-3 expression in odontoblast was restricted in mature odontoblasts beneath the cusps during the postnatal periods. This result was also supported by real-time RT-PCR analysis, and indicated that syndecan-3 may be involved in the progress of dentinogenesis rather than in the initiation of it. Syndecan-4 mRNA roughly showed comparable expression patterns to those of syndecan-3. Syndecan-2 mRNA did not show significant expression during the experimental period, but real-time RT-PCR analysis suggested that syndecan-2 expression might be enhanced with hard tissue formation.
Collapse
Affiliation(s)
- Kaoru Fujikawa
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo, Japan
| | - Naoko Nonaka
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo, Japan
| | - Xiaofang Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Shunichi Shibata
- Department of Maxillofacial Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. .,Department of Anatomy, School of Dentistry, Health Sciences University of Hokkaido, Tobetsu, Ishikari, Hokkaido, 061-0293, Japan.
| |
Collapse
|
41
|
Pérez LA, Leyton L, Valdivia A. Thy-1 (CD90), Integrins and Syndecan 4 are Key Regulators of Skin Wound Healing. Front Cell Dev Biol 2022; 10:810474. [PMID: 35186924 PMCID: PMC8851320 DOI: 10.3389/fcell.2022.810474] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Acute skin wound healing is a multistage process consisting of a plethora of tightly regulated signaling events in specialized cells. The Thy-1 (CD90) glycoprotein interacts with integrins and the heparan sulfate proteoglycan syndecan 4, generating a trimolecular complex that triggers bi-directional signaling to regulate diverse aspects of the wound healing process. These proteins can act either as ligands or receptors, and they are critical for the successful progression of wound healing. The expression of Thy-1, integrins, and syndecan 4 is controlled during the healing process, and the lack of expression of any of these proteins results in delayed wound healing. Here, we review and discuss the roles and regulatory events along the stages of wound healing that support the relevance of Thy-1, integrins, and syndecan 4 as crucial regulators of skin wound healing.
Collapse
Affiliation(s)
- Leonardo A. Pérez
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Program of Cellular & Molecular Biology, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
- Faculty of Medicine, Universidad de Chile, Santiago, Chile
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| | - Alejandra Valdivia
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, United States
- *Correspondence: Lisette Leyton, ; Alejandra Valdivia,
| |
Collapse
|
42
|
Pham SH, Pratt K, Okolicsanyi RK, Oikari LE, Yu C, Peall IW, Arif KMT, Chalmers TA, Gyimesi M, Griffiths LR, Haupt LM. Syndecan-1 and -4 influence Wnt signaling and cell migration in human breast cancers. Biochimie 2022; 198:60-75. [DOI: 10.1016/j.biochi.2022.01.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/30/2021] [Accepted: 01/21/2022] [Indexed: 11/16/2022]
|
43
|
The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev 2022; 181:114069. [PMID: 34838648 PMCID: PMC8860232 DOI: 10.1016/j.addr.2021.114069] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Comprehensive overview of different classes of ECM molecules in the HSC niche. Overview of current knowledge on role of biophysics of the HSC niche. Description of approaches to create artificial stem cell niches for several application. Importance of considering ECM in drug development and testing.
Hematopoietic stem cells (HSCs) are the life-long source of all types of blood cells. Their function is controlled by their direct microenvironment, the HSC niche in the bone marrow. Although the importance of the extracellular matrix (ECM) in the niche by orchestrating niche architecture and cellular function is widely acknowledged, it is still underexplored. In this review, we provide a comprehensive overview of the ECM in HSC niches. For this purpose, we first briefly outline HSC niche biology and then review the role of the different classes of ECM molecules in the niche one by one and how they are perceived by cells. Matrix remodeling and the emerging importance of biophysics in HSC niche function are discussed. Finally, the application of the current knowledge of ECM in the niche in form of artificial HSC niches for HSC expansion or targeted differentiation as well as drug testing is reviewed.
Collapse
|
44
|
Wen D, Cui J, Li P, Xiong Q, Chen G, Wu C. Syndecan-4 assists Mycobacterium tuberculosis entry into lung epithelial cells by regulating the Cdc42, N-WASP, and Arp2/3 signaling pathways. Microbes Infect 2022; 24:104931. [PMID: 35026388 DOI: 10.1016/j.micinf.2022.104931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 09/14/2021] [Accepted: 01/03/2022] [Indexed: 11/17/2022]
Abstract
Syndecan-4 (SDC4) is a transmembrane heparin sulfate proteoglycan that regulates inflammatory responses, cell motility, cell adhesion and intracellular signaling. In this study, we found that overexpression of SDC4 promoted the infection efficiency of Mycobacterium tuberculosis (Mtb), whereas knockdown of SDC4 reduced the infection efficiency, suggesting that SDC4 assisted Mtb infection of epithelial cells. We also observed that Mtb infection affected the F-actin/G-actin ratio, which was also correlated with SDC4 expression levels. Analysis of the Cdc42, N-WASP, and Arp2/3 signaling pathways during Mtb infection revealed that knockdown of Cdc42 and N-WASP or the addition of ZCL278, Wiskostatin or CK636 (blockers of Cdc42, N-WASP, and Arp2/3, respectively) significantly exacerbated Mtb infection in lung epithelial cells. Taken together, our data indicate that SDC4 assists Mtb infection of epithelial cells by regulating the Cdc42, N-WASP, and Arp2/3 signaling pathways, which regulate the polymerization of the actin cytoskeleton.
Collapse
Affiliation(s)
- Da Wen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Jia Cui
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; Department of Microbiology, Changzhi Medical College, Changzhi 046000, China
| | - Ping Li
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Qiuhong Xiong
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Guangxin Chen
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China.
| | - Changxin Wu
- Institute of Biomedical Sciences, Shanxi University, Taiyuan 030006, China; Key Lab of Medical Molecular Cell Biology of Shanxi Province, Shanxi University, Taiyuan 030006, China; The Provincial Key Laboratories for Prevention and Treatment of Major Infectious Diseases, Taiyuan 030006, Shanxi, China.
| |
Collapse
|
45
|
HIV-1 Tat and Heparan Sulfate Proteoglycans Orchestrate the Setup of in Cis and in Trans Cell-Surface Interactions Functional to Lymphocyte Trans-Endothelial Migration. Molecules 2021; 26:molecules26247488. [PMID: 34946571 PMCID: PMC8705413 DOI: 10.3390/molecules26247488] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 12/26/2022] Open
Abstract
HIV-1 transactivating factor Tat is released by infected cells. Extracellular Tat homodimerizes and engages several receptors, including integrins, vascular endothelial growth factor receptor 2 (VEGFR2) and heparan sulfate proteoglycan (HSPG) syndecan-1 expressed on various cells. By means of experimental cell models recapitulating the processes of lymphocyte trans-endothelial migration, here, we demonstrate that upon association with syndecan-1 expressed on lymphocytes, Tat triggers simultaneously the in cis activation of lymphocytes themselves and the in trans activation of endothelial cells (ECs). This "two-way" activation eventually induces lymphocyte adhesion and spreading onto the substrate and vascular endothelial (VE)-cadherin reorganization at the EC junctions, with consequent endothelial permeabilization, leading to an increased extravasation of Tat-presenting lymphocytes. By means of a panel of biochemical activation assays and specific synthetic inhibitors, we demonstrate that during the above-mentioned processes, syndecan-1, integrins, FAK, src and ERK1/2 engagement and activation are needed in the lymphocytes, while VEGFR2, integrin, src and ERK1/2 are needed in the endothelium. In conclusion, the Tat/syndecan-1 complex plays a central role in orchestrating the setup of the various in cis and in trans multimeric complexes at the EC/lymphocyte interface. Thus, by means of computational molecular modelling, docking and dynamics, we also provide a characterization at an atomic level of the binding modes of the Tat/heparin interaction, with heparin herein used as a structural analogue of the heparan sulfate chains of syndecan-1.
Collapse
|
46
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 507] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
47
|
Hu J, Li Y, Wei Z, Chen H, Sun X, Zhou Q, Zhang Q, Yin Y, Guo M, Chen J, Zhai G, Xu B, Xie J. A reduction in the vascular smooth muscle cell focal adhesion component syndecan-4 is associated with abdominal aortic aneurysm formation. Clin Transl Med 2021; 11:e605. [PMID: 34936241 PMCID: PMC8693440 DOI: 10.1002/ctm2.605] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a serious vascular disease for which there is no effective drug treatment. The incidence of AAA increases significantly as a subject ages, and the molecular mechanism of AAA formation remains elusive. In the present study, we investigated the role of syndecan-4 (SDC4), an important component of focal adhesions, in AAA formation and its association with phenotypic changes in vascular smooth muscle cells (VSMCs). METHODS AND RESULTS The protein expression levels of SDC4 were significantly decreased in human AAA tissue and those of an AAA mouse model. Moreover, SDC4 knockout (KO) in mice accelerated the formation and rupture of AAAs induced by angiotensin II (Ang II) and calcium chloride (CaCl2 ) Mechanistically, the decrease in SDC4 led to the transformation of cultured VSMCs from a contractile to a secretory phenotype. The RhoA-F/G-actin-myocardin-related transcription factor-A (MRTF-A) signalling pathway was shown to be involved in SDC4-dependent VSMC alteration. Sphingosine-1-phosphate (S1P), a G-protein-coupled receptor, attenuated the AAA formation in SDC4-KO and wild-type (WT) mice in response to Ang II and CaCl2 stimulation. CONCLUSION We herein demonstrated that silencing SDC4 was associated with increased AAA formation and phenotypic changes in VSMCs via the RhoA-F/G-actin-MRTF-A pathway. These findings indicated that a reduction in SDC4 expression was an important pathological alteration and potential therapeutic target for AAA formation.
Collapse
Affiliation(s)
- Jiaxin Hu
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Yuyu Li
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Haiting Chen
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Qing Zhou
- Department of Cardiac Surgery, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing UniversityNanjingChina
| | - Qi Zhang
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Yong Yin
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Meng Guo
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Jianzhou Chen
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Guangyao Zhai
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| | - Jun Xie
- Department of Cardiology, Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical School, MOE Key Laboratory of Model Animal for Disease Study, Nanjing UniversityNanjingChina
| |
Collapse
|
48
|
Biomechanical properties of endothelial glycocalyx: An imperfect pendulum. Matrix Biol Plus 2021; 12:100087. [PMID: 34820618 PMCID: PMC8596327 DOI: 10.1016/j.mbplus.2021.100087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/11/2021] [Accepted: 10/11/2021] [Indexed: 01/18/2023] Open
Abstract
This review seeks to fuse the discoveries in cell biology with mechanical engineering to produce a comprehensive biophysical model of endothelial glycocalyx. The aperiodic oscillatory motions of glycocalyx and cortical actin web underlie our prediction of two functional pacemakers and their participation in the outside-in signaling, the basis for mechanotransduction, and the dampening action of the inside-out signaling. Advancing an idea that the glycocalyx, plasma membrane, and cortical actin web represent a structure-functional unit and proposing the concept of tensegrity model. Presentation of our recent data suggesting that erythrocytes are gliding or havering and rotating over the surface of intact glycocalyx, whereas the rotational and hovering components of their passage along the capillaries are lost when glycocalyx of either is degraded.
Endothelial glycocalyx plays a crucial role in hemodynamics in health and disease, yet studying it is met by multiple technical hindrances. We attempted to outline our views on some biomechanical properties of endothelial glycocalyx, which are potentially amenable to mathematical modeling. We start with the null-hypothesis ascribing to glycocalyx the properties of a pendulum and reject this hypothesis on the grounds of multiple obstacles for pendulum behavior, such as rich decoration with flexible negatively charged side-chains, variable length and density, fluid fixation to the plasma membrane. We next analyze the current views on membrane attachments to the cortical actin web, its pulsatile contraction-relaxation cycles which rebound to the changes in tension of the plasma membrane. Based on this, we consider the outside-in signaling, the basis for mechanotransduction, and the dampening action of the inside-out signaling. The aperiodic oscillatory motions of glycocalyx and cortical actin web underlie our prediction of two functional pacemakers. We next advance an idea that the glycocalyx, plasma membrane, and cortical actin web represent a structure-functional unit and propose the concept of tensegrity model. Finally, we present our recent data suggesting that erythrocytes are gliding or hovering and rotating over the surface of intact glycocalyx, whereas the rotational and hovering components of their passage along the capillaries are lost when glycocalyx of either is degraded. These insights into the mechanics of endothelial glycocalyx motions may be of value in crosspollination between biomechanics, physiology, and pathophysiology for deeper appreciation of its rich untapped resources in health and pharmacotherapy in disease.
Collapse
|
49
|
Itoh Y. Modulation of Microenvironment Signals by Proteolytic Shedding of Cell Surface Extracellular Matrix Receptors. Front Cell Dev Biol 2021; 9:736735. [PMID: 34796172 PMCID: PMC8593224 DOI: 10.3389/fcell.2021.736735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/20/2021] [Indexed: 01/02/2023] Open
Abstract
Multicellular organisms are composed of cells and extracellular matrix (ECM). ECM is a network of multidomain macromolecules that fills gaps between cells. It acts as a glue to connect cells, provides scaffolding for migrating cells, and pools cytokines and growth factors. ECM also directly sends signals to the cells through ECM receptors, providing survival signals and migration cues. Altogether, ECM provides a correct microenvironment for the cells to function in the tissue. Although ECM acts as a signaling molecule, they are insoluble solid molecules, unlike soluble receptor ligands such as cytokines and growth factors. Upon cell binding to the ECM through ECM receptors and signals transmitted, cells then need to have a mechanism to release from ECM to prevent prolonged signals, which may be tumorigenic, and migrate on ECM. One effective means to release the cells from ECM is to cleave the ECM receptors by proteinases. In this mini-review, current knowledge of ECM receptor shedding will be discussed.
Collapse
Affiliation(s)
- Yoshifumi Itoh
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Moore KH, Murphy HA, Chapman H, George EM. Syncytialization alters the extracellular matrix and barrier function of placental trophoblasts. Am J Physiol Cell Physiol 2021; 321:C694-C703. [PMID: 34406903 DOI: 10.1152/ajpcell.00177.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The human placenta is of vital importance for proper nutrient and waste exchange, immune regulation, and overall fetal health and growth. Specifically, the extracellular matrix (ECM) of placental syncytiotrophoblasts, which extends outward from the placental chorionic villi into maternal blood, acts on a molecular level to regulate and maintain this barrier. Importantly, placental barrier dysfunction has been linked to diseases of pregnancy such as preeclampsia and intrauterine growth restriction. To help facilitate our understanding of the interface and develop therapeutics to repair or prevent dysfunction of the placental barrier, in vitro models of the placental ECM would be of great value. In this study, we aimed to characterize the ECM of an in vitro model of the placental barrier using syncytialized BeWo choriocarcinoma cells. Syncytialization caused a marked change in syndecans, integral proteoglycans of the ECM, which matched observations of in vivo placental ECM. Syndecan-1 expression increased greatly and predominated the other variants. Barrier function of the ECM, as measured by electric cell-substrate impedance sensing (ECIS), increased significantly during and after syncytialization, whereas the ability of THP-1 monocytes to adhere to syncytialized BeWos was greatly reduced compared with nonsyncytialized controls. Furthermore, ECIS measurements indicated that ECM degradation with matrix metalloproteinase-9 (MMP-9), but not heparanase, decreased barrier function. This decrease in ECIS-measured barrier function was not associated with any changes in THP-1 adherence to syncytialized BeWos treated with heparanase or MMP-9. Thus, syncytialization of BeWos provides a physiologically accurate placental ECM with a barrier function matching that seen in vivo.
Collapse
Affiliation(s)
- Kyle H Moore
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Haley A Murphy
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Heather Chapman
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Eric M George
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|